Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798688

ABSTRACT

Mutations in the human Ocular albinism type-1 gene OA1 are associated with abnormal retinal pigment epithelium (RPE) melanogenesis and poor binocular vision resulting from misrouting of ipsilateral retinal ganglion cell (iRGC) axons to the brain. We studied the latter using wild-type (WT) and Oa1-/- mouse eyes. At embryonic stages, the WT RPE-specific Oa1 protein signals through cAMP/Epac1-Erk2-CREB. Following CREB phosphorylation, a pCREB gradient extends from the RPE to the differentiating retinal amacrine and RGCs. In contrast to WT, the Oa1-/- RPE and ventral ciliary-margin-zone, a niche for iRGCs, express less pCREB while their retinas have a disrupted pCREB gradient, indicating Oa1's involvement in pCREB maintenance. Oa1-/- retinas also show hyperproliferation, enlarged nuclei, reduced differentiation, and fewer newborn amacrine and RGCs than WT retinas. Our results demonstrate that Oa1's absence leads to reduced binocular vision through a hyperproliferation-associated block in differentiation that impairs neurogenesis. This may affect iRGC axon's routing to the brain.

2.
PLoS One ; 13(3): e0194004, 2018.
Article in English | MEDLINE | ID: mdl-29538408

ABSTRACT

Extracellular vesicles (EVs) released by virtually every cell of all organisms are involved in processes of intercellular communication through the delivery of their functional mRNAs, proteins and bioactive lipids. We previously demonstrated that mouse embryonic stem cell-released EVs (mESEVs) are able to transfer their content to different target retinal cells, inducing morphological and biochemical changes in them. The main objective of this paper is to characterize EVs derived from human embryonic stem cells (hESEVs) and investigate the effects that they have on cultured retinal glial, progenitor Müller cells, which are known to give rise to retinal neurons under specific conditions. This would allow us to establish if hESEVs have a pro-regenerative potential not yet described that could be used in the future for treatment of human retinal degenerative diseases. Initially, we showed that hESEVs are heterogeneous in size, contain mRNAs and proteins involved in the induction and maintenance of stem cell pluripotency and can be internalized by cultured Müller cells. After a single exposure to hESEVs these cells display changes in their gene expression profile, and with multiple exposures they de-differentiate and trans-differentiate into retinal neuronal precursors. hESEVs were then fractionated into microvesicles (MVs) and exosomes (EXOs), which were characterized by size, specific surface proteins and biochemical/molecular components. We demonstrate that despite the similar internalization of non-fractionated hESEVs, MVs and EXOs by Müller progenitor cells, in vitro, only the release of MVs' cargo into the cells' cytoplasm induces specific changes in their levels of pluripotency mRNAs and early retinal proteins. EXOs do not produce any detectable effect. Thus, we conclude that MVs and MVs-containing hESEVs are promising agents that possibly could promote the regeneration of diseased or damaged retinas in vivo through inducing glial Müller cells to become replacement neurons.


Subject(s)
Ependymoglial Cells/physiology , Extracellular Vesicles/physiology , Human Embryonic Stem Cells/physiology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/physiology , Cells, Cultured , Ependymoglial Cells/metabolism , Exosomes/metabolism , Exosomes/physiology , Extracellular Vesicles/metabolism , HEK293 Cells , Human Embryonic Stem Cells/metabolism , Humans , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Neurons/physiology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , RNA, Messenger/metabolism , Regeneration/physiology , Retina/metabolism , Retina/physiology , Transcriptome/physiology
3.
PLoS One ; 11(9): e0162273, 2016.
Article in English | MEDLINE | ID: mdl-27607449

ABSTRACT

Ocular albinism type 1 (OA), caused by mutations in the OA1 gene, encodes a G-protein coupled receptor, OA1, localized in melanosomal membranes of the retinal pigment epithelium (RPE). This disorder is characterized by both RPE macro-melanosomes and abnormal decussation of ganglion cell axons at the brain's optic chiasm. We demonstrated previously that Oa1 specifically activates Gαi3, which also signals in the Oa1 transduction pathway that regulates melanosomal biogenesis. In this study, we screened the human Gαi3 gene, GNAI3, in DNA samples from 26 patients who had all clinical characteristics of OA but in whom a specific mutation in the OA1 gene had not been found, and in 6 normal control individuals. Using the Agilent HaloPlex Target Enrichment System and next-generation sequencing (NGS) on the Illumina MiSeq platform, we identified 518 variants after rigorous filtering. Many of these variants were corroborated by Sanger sequencing. Overall, 98.8% coverage of the GNAI3 gene was obtained by the HaloPlex amplicons. Of all variants, 6 non-synonymous and 3 synonymous were in exons, 41 in a non-coding exon embedded in the 3' untranslated region (UTR), 6 in the 5' UTR, and 462 in introns. These variants included novel SNVs, insertions, deletions, and a frameshift mutation. All were found in at least one patient but none in control samples. Using computational methods, we modeled the GNAI3 protein and its non-synonymous exonic mutations and determined that several of these may be the cause of disease in the patients studied. Thus, we have identified GNAI3 as a second gene possibly responsible for X-linked ocular albinism.


Subject(s)
Albinism, Ocular/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Genetic Association Studies , Genetic Predisposition to Disease , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Albinism, Ocular/diagnosis , Base Pairing/genetics , Base Sequence , Exons/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Genetic Loci , Heterozygote , Homozygote , Humans , Introns/genetics , Models, Molecular , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Reproducibility of Results , Sequence Analysis, DNA
4.
PLoS One ; 8(9): e76240, 2013.
Article in English | MEDLINE | ID: mdl-24098784

ABSTRACT

PURPOSE: Ocular Albinism type 1 (OA1) is a disease caused by mutations in the OA1 gene and characterized by the presence of macromelanosomes in the retinal pigment epithelium (RPE) as well as abnormal crossing of the optic axons at the optic chiasm. We showed in our previous studies in mice that Oa1 activates specifically Gαi3 in its signaling pathway and thus, hypothesized that a constitutively active Gαi3 in the RPE of Oa1-/- mice might keep on the Oa1 signaling cascade and prevent the formation of macromelanosomes. To test this hypothesis, we have generated transgenic mice that carry the constitutively active Gαi3 (Q204L) protein in the RPE of Oa1-/- mice and are now reporting the effects that the transgene produced on the Oa1-/- RPE phenotype. METHODS: Transgenic mice carrying RPE-specific expression of the constitutively active Gαi3 (Q204L) were generated by injecting fertilized eggs of Oa1-/- females with a lentivirus containing the Gαi3 (Q204L) cDNA. PCR, Southern blots, Western blots and confocal microscopy were used to confirm the presence of the transgene in the RPE of positive transgenic mice. Morphometrical analyses were performed using electron microscopy to compare the size and number of melanosomes per RPE area in putative Oa1-/-, Gαi3 (Q204L) transgenic mice with those of wild-type NCrl and Oa1-/- mice. RESULTS: We found a correlation between the presence of the constitutively active Gαi3 (Q204L) transgene and the rescue of the normal phenotype of RPE melanosomes in Oa1-/-, Gαi3 (Q204L) mice. These mice have higher density of melanosomes per RPE area and a larger number of small melanosomes than Oa1-/- mice, and their RPE phenotype is similar to that of wild-type mice. CONCLUSIONS: Our results show that a constitutively active Gαi3 protein can by-pass the lack of Oa1 protein in Oa1-/- mice and consequently rescue the RPE melanosomal phenotype.


Subject(s)
Albinism, Ocular/pathology , Eye Proteins/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Membrane Glycoproteins/genetics , Receptors, G-Protein-Coupled/genetics , Retinal Pigment Epithelium/abnormalities , Signal Transduction/physiology , Animals , Blotting, Southern , Blotting, Western , Female , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Membrane Glycoproteins/deficiency , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Receptors, G-Protein-Coupled/deficiency , Retinal Pigment Epithelium/metabolism
5.
J Biol Chem ; 288(14): 9742-9754, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23386608

ABSTRACT

The novel rhomboid-like protein RHBDD2 is distantly related to rhomboid proteins, a group of highly specialized membrane-bound proteases that catalyze regulated intramembrane proteolysis. In retina, RHBDD2 is expressed from embryonic stages to adulthood, and its levels show age-dependent changes. RHBDD2 is distinctly abundant in the perinuclear region of cells, and it localizes to their Golgi. A glycine zipper motif present in one of the transmembrane domains of RHBDD2 is important for its packing into the Golgi membranes. Its deletion causes dislodgment of RHBDD2 from the Golgi. A specific antibody against RHBDD2 recognizes two forms of the protein, one with low (39 kDa; RHBDD2(L)) and the other with high (117 kDa; RHBDD2H) molecular masses in mouse retinal extracts. RHBDD2(L) seems to be ubiquitously expressed in all retinal cells. In contrast, RHBDD2H seems to be present only in the outer segments of cone photoreceptors and may correspond to a homotrimer of RHBDD2(L). This protein consistently co-localizes with S- and M-types of cone opsins. We identified a homozygous mutation in the human RHBDD2 gene, R85H, that co-segregates with disease in affected members of a family with autosomal recessive retinitis pigmentosa. Our findings suggest that the RHBDD2 protein plays important roles in the development and normal function of the retina.


Subject(s)
Endopeptidases/biosynthesis , Endopeptidases/physiology , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Retina/metabolism , Retinitis Pigmentosa/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Female , Gene Expression Regulation , Gene Expression Regulation, Developmental , Glycine/chemistry , Golgi Apparatus/metabolism , HEK293 Cells , Homozygote , Humans , Immunohistochemistry/methods , In Situ Hybridization , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Opsins/chemistry , Pregnancy , Pregnancy, Animal , Retina/embryology , Retinal Cone Photoreceptor Cells/metabolism , Sequence Homology, Amino Acid
6.
PLoS One ; 6(9): e24376, 2011.
Article in English | MEDLINE | ID: mdl-21931697

ABSTRACT

BACKGROUND: Ocular albinism type 1, an X-linked disease characterized by the presence of enlarged melanosomes in the retinal pigment epithelium (RPE) and abnormal crossing of axons at the optic chiasm, is caused by mutations in the OA1 gene. The protein product of this gene is a G-protein-coupled receptor (GPCR) localized in RPE melanosomes. The Oa1-/- mouse model of ocular albinism reproduces the human disease. Oa1 has been shown to immunoprecipitate with the Gαi subunit of heterotrimeric G proteins from human skin melanocytes. However, the Gαi subfamily has three highly homologous members, Gαi1, Gαi2 and Gαi3 and it is possible that one or more of them partners with Oa1. We had previously shown by in-vivo studies that Gαi3-/- and Oa1-/- mice have similar RPE phenotype and decussation patterns. In this paper we analyze the specificity of the Oa1-Gαi interaction. METHODOLOGY: By using the genetic mouse models Gαi1-/-, Gαi2-/-, Gαi3-/- and the double knockout Gαi1-/-, Gαi3-/- that lack functional Gαi1, Gαi2, Gαi3, or both Gαi1 and Gαi3 proteins, respectively, we show that Gαi3 is critical for the maintenance of a normal melanosomal phenotype and that its absence is associated with changes in melanosomal size and density. GST-pull-down and immunoprecipitation assays conclusively demonstrate that Gαi3 is the only Gαi that binds to Oa1. Western blots show that Gαi3 expression is barely detectable in the Oa1-/- RPE, strongly supporting a previously unsuspected role for Gαi3 in melanosomal biogenesis. CONCLUSION: Our results identify the Oa1 transducer Gαi3 as the first downstream component in the Oa1 signaling pathway.


Subject(s)
Eye Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Melanosomes/metabolism , Membrane Glycoproteins/metabolism , Organelle Size , Receptors, G-Protein-Coupled/metabolism , Retinal Pigment Epithelium/metabolism , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Chromatography, Liquid , Electroretinography , Eye Proteins/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Humans , Mass Spectrometry , Melanosomes/ultrastructure , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Sequence Data , Organelle Shape , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/deficiency , Retinal Pigment Epithelium/physiology , Retinal Pigment Epithelium/ultrastructure
7.
Invest Ophthalmol Vis Sci ; 49(7): 3245-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18378571

ABSTRACT

PURPOSE: Ocular albinism type 1 (OA1) is characterized by abnormalities in retinal pigment epithelium (RPE) melanosomes and misrouting of optic axons. The OA1 gene encodes a G-protein-coupled receptor (GPCR) that coimmunoprecipitates with the G alpha i-subunit of heterotrimeric G-proteins from human melanocyte extracts. This study was undertaken to test whether one of the G alpha i proteins, G alpha i3, signals in the same pathway as OA1 to regulate melanosome biogenesis and axonal growth through the optic chiasm. METHODS: Adult G alpha i3(-/-) and Oa1(-/-) mice were compared with their respective control mice (129Sv and B6/NCrl) to study the effects of the loss of G alpha i3 or Oa1 function. Light and electron microscopy were used to analyze the morphology of the retina and the size and density of RPE melanosomes, electroretinograms to study retinal function, and retrograde labeling to investigate the size of the uncrossed optic pathway. RESULTS: Although G alpha i3(-/-) and Oa1(-/-) photoreceptors were comparable to those of the corresponding control retinas, the density of their RPE melanosomes was significantly lower than in control RPEs. In addition, the RPE cells of G alpha i3(-/-) and Oa1(-/-) mice showed abnormal melanosomes that were far larger than the largest 129Sv and B6/NCrl melanosomes, respectively. Although G alpha i3(-/-) and Oa1(-/-) mice had normal results on electroretinography, retrograde labeling showed a significant reduction from control in the size of their ipsilateral retinofugal projections. CONCLUSIONS: These results indicate that G alpha i3, like Oa1, plays an important role in melanosome biogenesis. Furthermore, they suggest a common Oa1-G alpha i3 signaling pathway that ultimately affects axonal growth through the optic chiasm.


Subject(s)
Eye Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Melanosomes/physiology , Membrane Glycoproteins/metabolism , Optic Chiasm/physiology , Pigment Epithelium of Eye/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Axons/physiology , Carrier Proteins/metabolism , Cell Count , Electroretinography , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Intracellular Membranes/metabolism , Melanosomes/ultrastructure , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout , Microscopy, Electron , Phenotype , Pigment Epithelium of Eye/ultrastructure , Receptors, G-Protein-Coupled/deficiency , Retina/cytology , Retina/physiology , Retinal Ganglion Cells/cytology , Signal Transduction/physiology , Synaptic Transmission , Visual Pathways/cytology , Visual Pathways/growth & development , Visual Pathways/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...